1
|
Kotb MA, Abdelmawgood IA, Ibrahim IM. Pharmacophore-based virtual screening, molecular docking, and molecular dynamics investigation for the identification of novel, marine aromatase inhibitors. BMC Chem 2024; 18:235. [PMID: 39593184 PMCID: PMC11590544 DOI: 10.1186/s13065-024-01350-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Breast cancer remains a leading cause of mortality among women worldwide. Our current research focuses on identifying effective therapeutic agents by targeting the human aromatase enzyme. Aromatase inhibitors (AIs) have been effective in treating postmenopausal breast cancer but face challenges such as drug resistance and long-term side effects like cognitive decline and osteoporosis. Natural products, especially from marine organisms, are emerging as potential sources for new drug candidates due to their structural diversity and pharmacological properties. This study aims to discover marine natural products capable of inhibiting human aromatase by combining ligand-based and structure-based pharmacophore models for virtual screening against the Comprehensive Marine Natural Products Database. From the initial virtual screening of more than 31,000 compounds, 1,385 marine natural products were identified as possible candidates. Following initial molecular docking analysis, only four compounds managed to pass the criteria this research has introduced to confirm strong binding affinity to aromatase. All four compounds yielded acceptable binding affinities, with CMPND 27987 having the highest -10.1 kcal/mol. All four hits were subjected to molecular dynamics, and CMPND 27987 was further confirmed to be the most stable at the protein's active site, with an MM-GBSA free binding energy of -27.75 kcal/mol. Our in silico studies indicate that CMPND 27987 interacts effectively within the binding site of the human aromatase, maintaining high affinity and stability. Based on these findings, we propose that CMPND 27987 could hold significant potential for further lead optimization and drug development.
Collapse
Affiliation(s)
- Mohamed A Kotb
- 1Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | | | - Ibrahim M Ibrahim
- 2Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
2
|
Bae E, Beil S, König M, Stolte S, Escher BI, Markiewicz M. The mode of toxic action of ionic liquids: Narrowing down possibilities using high-throughput, in vitro cell-based bioassays. ENVIRONMENT INTERNATIONAL 2024; 193:109089. [PMID: 39500119 DOI: 10.1016/j.envint.2024.109089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 11/25/2024]
Abstract
Growing concerns about the environmental impact of ionic liquids (ILs) have spurred research into their (eco)toxic effects, but studies on their mode of toxic action (MOA) still remain limited. However, understanding the MOA and identifying structural features responsible for enhanced toxicity is crucial for characterising the hazard and designing safer alternatives. Therefore, 45 ILs, with systematically varied chemical structures, were tested for cytotoxicity and two specific endpoints in reporter gene assays targeting the Nrf2-ARE mediated oxidative stress response (AREc32) and aryl hydrocarbon receptor activation (AhR-CALUX). While none of the ILs activated the reporter genes, cytotoxicity was high and markedly different between cell lines. Seven and 25 ILs proved more cytotoxic than predicted by baseline toxicity model in the AREc32 and the AhR-CALUX assays, respectively. The length of the side chain and headgroup structures of ILs altered the MOA of ILs. Cellular metabolism of the ILs, investigated by LC-MS/MS, showed side-chain oxidation of the long-chain quaternary ammonium compounds in AhR-CALUX cells and, to a lower extent, in AREc32 cells, however, this transformation could not explain the high cytotoxicity. Effect data for 72 ILs for ten endpoints retrieved from the Tox21 database identified the inhibition of aromatase activity and of mitochondrial membrane potential as potential MOAs. However, in vitro fluorimetric assays for these endpoints demonstrated that effects were activated in a non-specific manner, probably through cytotoxicity. Although many of the ILs tested induced cytotoxicity at concentrations lower than baseline toxicity, the specific MOAs responsible could not be identified. Alternatively, we suggest that the descriptors currently used may fail to define the affinity of ILs for cells. Testing of the affinity of ILs for a diverse range of biomolecules is needed to accurately describe their interactions with cells.
Collapse
Affiliation(s)
- Eunhye Bae
- Institute of Water Chemistry, Dresden University of Technology, D-01062 Dresden, Germany
| | - Stephan Beil
- Institute of Water Chemistry, Dresden University of Technology, D-01062 Dresden, Germany
| | - Maria König
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research-UFZ, D-04318 Leipzig, Germany
| | - Stefan Stolte
- Institute of Water Chemistry, Dresden University of Technology, D-01062 Dresden, Germany
| | - Beate I Escher
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research-UFZ, D-04318 Leipzig, Germany; Environmental Toxicology, Department of Geosciences, Eberhard Karls University Tübingen, D-72076 Tübingen, Germany
| | - Marta Markiewicz
- Institute of Water Chemistry, Dresden University of Technology, D-01062 Dresden, Germany.
| |
Collapse
|
3
|
Gheidari D, Mehrdad M, Hoseini F. Virtual screening, molecular docking, MD simulation studies, DFT calculations, ADMET, and drug likeness of Diaza-adamantane as potential MAPK ERK inhibitors. Front Pharmacol 2024; 15:1360226. [PMID: 39021828 PMCID: PMC11253198 DOI: 10.3389/fphar.2024.1360226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/07/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction: Multiple sclerosis (MS) is an autoimmune and inflammatory disease that destroys the protective coating of central nervous system (CNS) nerve fibers and affects over 2.8 million people worldwide. Despite several studies on new therapeutic targets and lead compounds, MS disease has limited treatment options. This condition may be caused by a complicated interaction of environmental and genetic variables. Studies showed that MS-associated microglial cells' increased MAPKERK activity may cause CNS inflammation and oligodendrocyte damage. Thus, screening for lead compounds that inhibit MAPKERK may protect brain cells and slow disease progression. Methods: The study aims to discover compounds that may inhibit MAPKERK as a novel approach for protecting the nervous system in managing MS. The study includes in silico methods, such as virtual screening, molecular docking, Density-functional theory (DFT) investigations (using the B3LYP/6-31++G(d,p) basis set in a gas phase environment), drug likeness scores, and molecular dynamic (MD) simulations. Results and Discussion:During the docking process with the MAPKERK protein, it was shown that the ligand L12 receptor had the best binding affinity, with a docking score of -6.18 kcal/mol. To investigate the stability of the binding, a 100 ns MD simulation was performed on the complex formed by the MAPKERK protein and L12. The receptor-ligand combination exhibited significant stability throughout the duration of the MD simulation. Additionally, the pharmacokinetic and drug-likeness properties of these ligands suggest that they have the potential to be considered viable candidates for future development in MS management.
Collapse
Affiliation(s)
- Davood Gheidari
- Department of Chemistry, Faculty of Science, University of Guilan, Rasht, Iran
| | - Morteza Mehrdad
- Department of Chemistry, Faculty of Science, University of Guilan, Rasht, Iran
| | | |
Collapse
|
4
|
Pasha MH, Gondal HY, Munir S, Alhussain SA, Zaki MEA. New enantioenriched β-indolyl ketones as aromatase inhibitors: Unraveling heme-ligand interactions by MD simulation and MMPBSA analysis. Arch Pharm (Weinheim) 2024; 357:e2400010. [PMID: 38578079 DOI: 10.1002/ardp.202400010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
A series of enantioenriched β-indolyl ketones as aromatase inhibitors (AI) is synthesized through the Michael-type Friedel-Crafts alkylation of indole. A highly efficient bifunctionalized amino catalyst is developed to access structurally diverse β-indolyl ketones in high yields (up to 91%) and excellent enantioselectivity (enantiomeric ratio up to 98:2). All the synthesized compounds demonstrated promising aromatase inhibitory potential, where ortho-substituted analogs (3c and 3e) were found most active with IC50 values of 0.68 and 0.90 µM, respectively. Both of these compounds exhibited significant cytotoxicity (IC50 = 0.34 and 0.37 µM) against the MCF-7 breast cancer cell line in the (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide) assay. Molecular docking studies of the synthesized compounds demonstrate favorable binding interactions with the estrogens controlling CYP19A1 (3EQM) and metabolizing CYP3A4 (5VCC) enzymes. Molecular dynamic (MD) simulation analysis revealed the essentiality of heme-ligand interactions to build a stable protein-ligand complex. An average root mean square deviation of 0.35 nm observed during a 100-ns MD simulation and binding free energy in the range of -190 to -227 kJ/mol calculated by g_mmpbsa analysis authenticated the stability of the 3c-3EQM complex. ADMET and drug-likeness parameters supported the suitability of these indole derivatives as the drug lead to develop potent inhibitors for estrogen-dependent breast cancer.
Collapse
Affiliation(s)
- Maira Hasnain Pasha
- Institute of Chemistry, Faculty of Science, University of Sargodha, Sargodha, Pakistan
| | | | - Shanza Munir
- Institute of Chemistry, Faculty of Science, University of Sargodha, Sargodha, Pakistan
| | - Sami A Alhussain
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Magdi E A Zaki
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Maghraby MTE, Mazyad Almutairi T, Bräse S, Salem OIA, Youssif BGM, Sheha MM. New 1,2,3-Triazole/1,2,4-triazole Hybrids as Aromatase Inhibitors: Design, Synthesis, and Apoptotic Antiproliferative Activity. Molecules 2023; 28:7092. [PMID: 37894571 PMCID: PMC10609154 DOI: 10.3390/molecules28207092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/29/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
A novel series of 1,2,3-triazole/1,2,4-triazole hybrids 5a, 5b, and 6a-i was designed and synthesized as antiproliferative agents targeting aromatase enzymes. The antiproliferative activity of the new hybrids against four cancer cells was studied using Erlotinib as a control. Compounds 6a and 6b demonstrated the highest antiproliferative activity among these hybrids, with GI50 values of 40 nM and 35 nM, respectively. Compound 6b was the most potent derivative, with a GI50 of 35 nM, comparable to Erlotinib's GI50 of 33 nM. Compound 6b inhibited all cancer cell lines with comparable efficacy to Erlotinib. Compounds 5a, 5b, and 6a-i were tested for inhibitory action against aromatase as a potential target for their antiproliferative activity. Results revealed that compounds 6a and 6b were the most potent aromatase inhibitors, with IC50 values of 0.12 ± 0.01 µM and 0.09 ± 0.01 µM, respectively, being more potent than the reference Ketoconazole (IC50 = 2.6 ± 0.20 µM) but less potent than Letrozole (IC50 = 0.002 ± 0.0002). These findings indicated that compounds 6a and 6b had significant aromatase inhibitory action and are potential antiproliferative candidates. The findings were further linked to molecular docking investigations, which gave models of strong interactions with the aromatase domain for inhibitors with high binding scores.
Collapse
Affiliation(s)
- Mohamed T-E Maghraby
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt; (M.T.-E.M.); (O.I.A.S.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, New Valley University, New Valley 72511, Egypt
| | - Tahani Mazyad Almutairi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Ola I. A. Salem
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt; (M.T.-E.M.); (O.I.A.S.)
| | - Bahaa G. M. Youssif
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt; (M.T.-E.M.); (O.I.A.S.)
| | - Mahmoud M. Sheha
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt;
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sphinx University, New-Assiut 71684, Egypt
| |
Collapse
|
6
|
Cobos-Ontiveros LA, Romero-Hernández LL, Mastranzo-Sánchez EB, Colín-Lozano B, Puerta A, Padrón JM, Merino-Montiel P, Vega Baez JL, Montiel-Smith S. Synthesis, antiproliferative evaluation and in silico studies of a novel steroidal spiro morpholinone. Steroids 2023; 192:109173. [PMID: 36621620 DOI: 10.1016/j.steroids.2023.109173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/07/2023]
Abstract
Estrogens play a pivotal role in the development of estrogen-dependent breast cancer and other hormone-dependent disorders. A common strategy to overcome the pathological effects of estrogens is the use of aromatase inhibitors (AIs), which bind to the enzyme and prevent the union with the natural substrate, decreasing the amount of estrogens produced. Several AIs have been developed, including inhibitors with a steroidal backbone and a nitrogen heterocycle in their structure. Encouraged by the notable results presented by current and clinical steroidal drugs, herein we present the synthesis of a steroidal spiro morpholinone derivative as a plausible aromatase inhibitor. The morpholinone derivative was synthesized over a six-step methodology starting from estrone. The title compound and its hydroxychloroacetamide derivative precursor were evaluated for their antiproliferative profile against estrogen-dependent and independent solid tumor cell lines: A549, HBL-100, HeLa, SW1573, T-47D and WiDr. Both compounds exhibited a potent antiproliferative activity in the micromolar range against the six cancer cell lines, with the hydroxychloroacetamide derivative precursor being a more potent inhibitor (GI50 = 0.25-2.4 µM) than the morpholinone derivative (GI50 = 2.0-11 µM). Furthermore, both compounds showed, in almost all cases, better GI50 values than the steroidal anticancer drugs abiraterone and galeterone. Docking simulations of the derivatives were performed in order to explain the experimental biological activity. The results showed interactions with the iron heme (derivative 3) and important residues of the steroidal binding-site (Met374) for the inhibition of human aromatase. A correlation was found between in vitro assays and the score obtained from the molecular docking study.
Collapse
Affiliation(s)
- Luis A Cobos-Ontiveros
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico
| | - Laura L Romero-Hernández
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico.
| | - Eduardo B Mastranzo-Sánchez
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico
| | - Blanca Colín-Lozano
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Universidad de La Laguna, c/ Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Universidad de La Laguna, c/ Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| | - Penélope Merino-Montiel
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico.
| | - Jose Luis Vega Baez
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico
| | - Sara Montiel-Smith
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico
| |
Collapse
|
7
|
Osmaniye D, Hıdır A, Sağlık BN, Levent S, Özkay Y, Kaplancıklı ZA. Synthesis of New Pyrimidine-Triazole Derivatives and Investigation of Their Anticancer Activities. Chem Biodivers 2022; 19:e202200216. [PMID: 35699405 DOI: 10.1002/cbdv.202200216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/13/2022] [Indexed: 11/09/2022]
Abstract
Aromatase inhibitors are the most used anticancer drug group in breast cancer cases. The development of resistance in cancer patients over time and the side effects of existing drugs make the need for new and effective agents permanent. In this study, 10 novel pyrimidine-triazole derivatives were synthesized and their anticancer activities were investigated. Compounds 5c and 5g showed inhibitor activity against MCF-7 cell line with IC 50 =1.573±0.020; 3.698±0.056 µM value, respectively. As a result of in vitro aromatase enzyme inhibition test, compounds 5c and 5g were exhibited significant activity with IC 50 =0.082±0.007 µM and IC50=0.198±0.015 µM, respectively. Estimated physicochemical parameters were calculated using the online SwissADME program for all compounds. Interaction modes of the compounds 5c and 5g were investigated against aromatase enzyme by means of docking studies. As a result of the studies, the importance of the triazole ring for aromatase inhibition has been understood.
Collapse
Affiliation(s)
- Derya Osmaniye
- Anadolu Universitesi, Pharmaceutical Chemistry, Anadolu University Faculty of Pharmacy Pharmaceutical Department, 26470, Eskisehir, TURKEY
| | - Arzu Hıdır
- Anadolu University: Anadolu Universitesi, Pharmaceutical Chemistry, Yunus Emre Campus, Eskişehir, TURKEY
| | - Begüm Nurpelin Sağlık
- Anadolu University: Anadolu Universitesi, Pharmaceutical Chemistry, Yunus Emre Campus, Eskişehir, TURKEY
| | - Serkan Levent
- Anadolu University: Anadolu Universitesi, Pharmaceutical Chemistry, Yunus Emre Campus, Eskişehir, TURKEY
| | - Yusuf Özkay
- Anadolu University: Anadolu Universitesi, Pharmaceutical Chemistry, Yunus Emre Campus, Eskişehir, TURKEY
| | - Zafer Asım Kaplancıklı
- Anadolu University: Anadolu Universitesi, Pharmaceutical Chemistry, Yunus Emre Campus, Eskişehir, TURKEY
| |
Collapse
|
8
|
Design, Semisynthesis, and Estrogenic Activity of Lignan Derivatives from Natural Dibenzylbutyrolactones. Pharmaceuticals (Basel) 2022; 15:ph15050585. [PMID: 35631411 PMCID: PMC9145393 DOI: 10.3390/ph15050585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 12/25/2022] Open
Abstract
Based on molecular docking studies on the ERα, a series of lignan derivatives (3–16) were designed and semisynthesized from the natural dibenzylbutyrolactones bursehernin (1) and matairesinol dimethyl ether (2). To examine their estrogenic and antiestrogenic potencies, the effects of these compounds on estrogen receptor element (ERE)-driven reporter gene expression and viability in human ER+ breast cancer cells were evaluated. Lignan compounds induced ERE-driven reporter gene expression with very low potency as compared with the pure agonist E2. However, coincubation of 5 μM of lignan derivatives 1, 3, 4, 7, 8, 9, 11, 13, and 14 with increasing concentrations of E2 (from 0.01 pM to 1 nM) reduced both the potency and efficacy of pure agonists. The binding to the rhERα-LBD was validated by TR-FRET competitive binding assay and lignans bound to the rhERα with IC50 values from 0.16 μM (compound 14) to 6 μM (compound 4). Induced fit docking (IFD) and molecular dynamics (MD) simulations for compound 14 were carried out to further investigate the binding mode interactions. Finally, the in silico ADME predictions indicated that the most potent lignan derivatives exhibited good drug-likeness.
Collapse
|
9
|
Design and synthesis of 6-amino-quinoxaline-alkynyl as potential aromatase (CYP19A1) inhibitors. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
10
|
Caballero Alfonso AY, Mora Lagares L, Novic M, Benfenati E, Kumar A. Exploration of structural requirements for azole chemicals towards human aromatase CYP19A1 activity: Classification modeling, structure-activity relationships and read-across study. Toxicol In Vitro 2022; 81:105332. [PMID: 35176449 DOI: 10.1016/j.tiv.2022.105332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/10/2022] [Accepted: 02/10/2022] [Indexed: 01/23/2023]
Abstract
Human aromatase, also called CYP19A1, plays a major role in the conversion of androgens into estrogens. Inhibition of aromatase is an important target for estrogen receptor (ER)-responsive breast cancer therapy. Use of azole compounds as aromatase inhibitors is widespread despite their low selectivity. A toxicological evaluation of commonly used azole-based drugs and agrochemicals with respect to CYP19A1is currently requested by the European Union- Registration, Evaluation, Authorization and Restriction of Chemicals (EU-REACH) regulations due to their potential as endocrine disruptors. In this connection, identification of structural alerts (SAs) is an effective strategy for the toxicological assessment and safe drug design. The present study describes the identification of SAs of azole-based chemicals as guiding experts to predict the aromatase activity. Total 21 SAs associated with aromatase activity were extracted from dataset of 326 azole-based drugs/chemicals obtained from Tox21 library. A cross-validated classification model having high accuracy (error rate 5%) was proposed which can precisely classify azole chemicals into active/inactive toward aromatase. In addition, mechanistic details and toxicological properties (agonism/antagonism) of azoles with respect to aromatase were explored by comparing active and inactive chemicals using structure-activity relationships (SAR). Lastly, few structural alerts were applied to form chemical categories for read-across applications.
Collapse
Affiliation(s)
- Ana Y Caballero Alfonso
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di RicercheFarmacologiche "Mario Negri"-IRCCS, Milano, Italy; Jozef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia
| | - Liadys Mora Lagares
- Jozef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia; Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, Ljubljana, Slovenia
| | - Marjana Novic
- Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, Ljubljana, Slovenia
| | - Emilio Benfenati
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di RicercheFarmacologiche "Mario Negri"-IRCCS, Milano, Italy
| | - Anil Kumar
- Department of Applied Sciences, University Institute of Engineering and Technology, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
11
|
Kumar D, Sharma P, Mahajan A, Dhawan R, Dua K. Pharmaceutical interest of in-silico approaches. PHYSICAL SCIENCES REVIEWS 2022. [DOI: 10.1515/psr-2018-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The virtual environment within the computer using software performed on the computer is known as in-silico studies. These drugs designing software play a vital task in discovering new drugs in the field of pharmaceuticals. These designing programs and software are employed in gene sequencing, molecular modeling, and in assessing the three-dimensional structure of the molecule, which can further be used in drug designing and development. Drug development and discovery is not only a powerful, extensive, and an interdisciplinary system but also a very complex and time-consuming method. This book chapter mainly focused on different types of in-silico approaches along with their pharmaceutical applications in numerous diseases.
Collapse
Affiliation(s)
- Dinesh Kumar
- Sri Sai College of Pharmacy , Manawala , Amritsar 143001 , Punjab , India
| | - Pooja Sharma
- Department of Pharmaceutical Sciences and Drug Research , Punjabi University , Patiala 147002 , Punjab , India
- Khalsa College of Pharmacy , Amritsar 143001 , Punjab , India
| | - Ayush Mahajan
- Sri Sai College of Pharmacy , Manawala , Amritsar 143001 , Punjab , India
| | - Ravi Dhawan
- Khalsa College of Pharmacy , Amritsar 143001 , Punjab , India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney , Ultimo 2007 , NSW , Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney , Ultimo 2007 , New South Wales , Australia
| |
Collapse
|
12
|
Maccallini C, Gallorini M, Sisto F, Akdemir A, Ammazzalorso A, De Filippis B, Fantacuzzi M, Giampietro L, Carradori S, Cataldi A, Amoroso R. New azolyl-derivatives as multitargeting agents against breast cancer and fungal infections: synthesis, biological evaluation and docking study. J Enzyme Inhib Med Chem 2021; 36:1632-1645. [PMID: 34289751 PMCID: PMC8300937 DOI: 10.1080/14756366.2021.1954918] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 01/06/2023] Open
Abstract
Nonsteroidal aromatase inhibitors (NSAIs) are well-established drugs for the therapy of breast cancer. However, they display some serious side effects, and their efficacy can be compromised by the development of chemoresistance. Previously, we have reported different indazole-based carbamates and piperidine-sulphonamides as potent aromatase inhibitors. Starting from the most promising compounds, here we have synthesised new indazole and triazole derivatives and evaluated their biological activity as potential dual agents, targeting both the aromatase and the inducible nitric oxide synthase, being this last dysregulated in breast cancer. Furthermore, selected compounds were evaluated as antiproliferative and cytotoxic agents in the MCF-7 cell line. Moreover, considering the therapeutic diversity of azole-based compounds, all the synthesized compounds were also evaluated as antifungals on different Candida strains. A docking study, as well as molecular dynamics simulation, were carried out to shed light on the binding mode of the most interesting compound into the different target enzymes catalytic sites.
Collapse
Affiliation(s)
- Cristina Maccallini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| | - Marialucia Gallorini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| | - Francesca Sisto
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Atilla Akdemir
- Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakif University, Computer-aided drug discovery laboratory, Istanbul, Turkey
| | | | - Barbara De Filippis
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| | | | - Letizia Giampietro
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| | - Simone Carradori
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
- Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakif University, Computer-aided drug discovery laboratory, Istanbul, Turkey
| | - Amelia Cataldi
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| | - Rosa Amoroso
- Department of Pharmacy, University “G. d’Annunzio” of Chieti -Pescara, Chieti, Italy
| |
Collapse
|
13
|
Giampietro L, Gallorini M, Gambacorta N, Ammazzalorso A, De Filippis B, Della Valle A, Fantacuzzi M, Maccallini C, Mollica A, Cataldi A, Nicolotti O, Amoroso R. Synthesis, structure-activity relationships and molecular docking studies of phenyldiazenyl sulfonamides as aromatase inhibitors. Eur J Med Chem 2021; 224:113737. [PMID: 34365129 DOI: 10.1016/j.ejmech.2021.113737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/29/2021] [Accepted: 08/01/2021] [Indexed: 12/11/2022]
Abstract
The exploration of innovative aromatase inhibitors represents an important approach for the identification of new therapeutic treatments of breast cancer. In this respect, a series of phenyldiazenyl sulfonamides was designed, synthesized and tested. Compounds 3b, 3f and 5f showed an aromatase inhibition in the micromolar range and were evaluated in vitro on the human breast cancer cell line MCF7 by MTT assay, cytotoxicity assay (LDH release), cell cycle analysis and apoptosis, revealing a dose-dependent inhibition profile. In particular, 3f displayed the best reduction in terms of metabolic activity and an anti-proliferative effect on MCF7 cells, being blocked in the G1/S phase checkpoint. Moreover, computational studies were carried out to better understand at a molecular level of detail the rationale behind the effective binding to the active site of aromatase of the more active inhibitor 3f. The obtained results allow to consider this compound as an interesting lead for the development of a new class of non-steroidal aromatase inhibitors.
Collapse
Affiliation(s)
- Letizia Giampietro
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. d'Annunzio" University, Chieti, Italy.
| | - Marialucia Gallorini
- Unit of Anatomy, Department of Pharmacy, "G. d'Annunzio" University, Chieti, Italy
| | - Nicola Gambacorta
- Department of Pharmacy-Drug Science, University of Bari "Aldo Moro", Via E. Orabona, 4, 70126, Bari, Italy
| | - Alessandra Ammazzalorso
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. d'Annunzio" University, Chieti, Italy
| | - Barbara De Filippis
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. d'Annunzio" University, Chieti, Italy
| | - Alice Della Valle
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. d'Annunzio" University, Chieti, Italy
| | - Marialuigia Fantacuzzi
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. d'Annunzio" University, Chieti, Italy
| | - Cristina Maccallini
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. d'Annunzio" University, Chieti, Italy
| | - Adriano Mollica
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. d'Annunzio" University, Chieti, Italy
| | - Amelia Cataldi
- Unit of Anatomy, Department of Pharmacy, "G. d'Annunzio" University, Chieti, Italy
| | - Orazio Nicolotti
- Department of Pharmacy-Drug Science, University of Bari "Aldo Moro", Via E. Orabona, 4, 70126, Bari, Italy
| | - Rosa Amoroso
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. d'Annunzio" University, Chieti, Italy
| |
Collapse
|
14
|
Bolnykh V, Rossetti G, Rothlisberger U, Carloni P. Expanding the boundaries of ligand–target modeling by exascale calculations. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2021. [DOI: 10.1002/wcms.1535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Viacheslav Bolnykh
- Laboratory of Computational Chemistry and Biochemistry École Polytechnique Fédérale de Lausanne Lausanne Switzerland
- Computational Biomedicine, Institute of Neuroscience and Medicine (INM‐9)/Institute for Advanced Simulations (IAS‐5) Forschungszentrum Jülich Jülich Germany
| | - Giulia Rossetti
- Computational Biomedicine, Institute of Neuroscience and Medicine (INM‐9)/Institute for Advanced Simulations (IAS‐5) Forschungszentrum Jülich Jülich Germany
- Jülich Supercomputing Centre (JSC) Forschungszentrum Jülich Jülich Germany
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation University Hospital Aachen RWTH Aachen University Aachen Germany
| | - Ursula Rothlisberger
- Laboratory of Computational Chemistry and Biochemistry École Polytechnique Fédérale de Lausanne Lausanne Switzerland
| | - Paolo Carloni
- Institute for Neuroscience and Medicine and Institute for Advanced Simulations (IAS‐5/INM‐9) “Computational Biomedicine” Forschungszentrum Jülich Jülich Germany
- JARA‐Institute INM‐11 “Molecular Neuroscience and Neuroimaging” Forschungszentrum Jülich Jülich Germany
| |
Collapse
|
15
|
Alhadrami HA, Sayed AM, Melebari SA, Khogeer AA, Abdulaal WH, Al-Fageeh MB, Algahtani M, Rateb ME. Targeting allosteric sites of human aromatase: a comprehensive in-silico and in-vitro workflow to find potential plant-based anti-breast cancer therapeutics. J Enzyme Inhib Med Chem 2021; 36:1334-1345. [PMID: 34139914 PMCID: PMC8759730 DOI: 10.1080/14756366.2021.1937145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recent findings suggested several allosteric pockets on human aromatase that could be utilised for the development of new modulators able to inhibit this enzyme in a new mechanism. Herein, we applied an integrated in-silico-based approach supported by in-vitro enzyme-based and cell-based validation assays to select the best leads able to target these allosteric binding sites from a small library of plant-derived natural products. Chrysin, apigenin, and resveratrol were found to be the best inhibitors targeting the enzyme’s substrate access channel and were able to produce a competitive inhibition with IC50 values ranged from 1.7 to 15.8 µM. Moreover, they showed a more potent antiproliferative effect against ER+ (MCF-7) than ER- one (MDA-MB-231) cell lines. On the other hand, both pomiferin and berberine were the best hits for the enzyme’s haem-proximal cavity producing a non-competitive inhibition (IC50 15.1 and 21.4 µM, respectively) and showed selective antiproliferative activity towards MCF-7 cell lines.
Collapse
Affiliation(s)
- Hani A Alhadrami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Molecular Diagnostic Lab, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah, Saudi Arabia.,Molecular Diagnostic Unit, The Regional Laboratory in Makkah, Ministry of Health, Makkah, Kingdom of Saudi Arabia
| | - Ahmed M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni Suef, Egypt
| | - Sami A Melebari
- Molecular Diagnostic Unit, The Regional Laboratory in Makkah, Ministry of Health, Makkah, Kingdom of Saudi Arabia
| | - Asem A Khogeer
- Plan and Research Department, General Directorate of Health Affairs, Makkah region, Ministry of Health, Makkah, Kingdom of Saudi Arabia
| | - Wesam H Abdulaal
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed B Al-Fageeh
- General Directorate for Funds and Grants (GDFG), King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Mohammad Algahtani
- Department of Laboratory and Blood Bank, Security Forces Hospital Program, Mecca, Saudi Arabia
| | - Mostafa E Rateb
- School of Computing, Engineering & Physical Sciences, University of the West of Scotland, Paisley, UK
| |
Collapse
|
16
|
Varela‐Rial A, Majewski M, De Fabritiis G. Structure based virtual screening: Fast and slow. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2021. [DOI: 10.1002/wcms.1544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Alejandro Varela‐Rial
- Acellera Labs Barcelona Spain
- Computational Science Laboratory Universitat Pompeu Fabra, Barcelona Biomedical Research Park (PRBB) Barcelona Spain
| | - Maciej Majewski
- Computational Science Laboratory Universitat Pompeu Fabra, Barcelona Biomedical Research Park (PRBB) Barcelona Spain
| | - Gianni De Fabritiis
- Computational Science Laboratory Universitat Pompeu Fabra, Barcelona Biomedical Research Park (PRBB) Barcelona Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA) Barcelona Spain
| |
Collapse
|
17
|
Spinello A, Borišek J, Pavlin M, Janoš P, Magistrato A. Computing Metal-Binding Proteins for Therapeutic Benefit. ChemMedChem 2021; 16:2034-2049. [PMID: 33740297 DOI: 10.1002/cmdc.202100109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Indexed: 01/18/2023]
Abstract
Over one third of biomolecules rely on metal ions to exert their cellular functions. Metal ions can play a structural role by stabilizing the structure of biomolecules, a functional role by promoting a wide variety of biochemical reactions, and a regulatory role by acting as messengers upon binding to proteins regulating cellular metal-homeostasis. These diverse roles in biology ascribe critical implications to metal-binding proteins in the onset of many diseases. Hence, it is of utmost importance to exhaustively unlock the different mechanistic facets of metal-binding proteins and to harness this knowledge to rationally devise novel therapeutic strategies to prevent or cure pathological states associated with metal-dependent cellular dysfunctions. In this compendium, we illustrate how the use of a computational arsenal based on docking, classical, and quantum-classical molecular dynamics simulations can contribute to extricate the minutiae of the catalytic, transport, and inhibition mechanisms of metal-binding proteins at the atomic level. This knowledge represents a fertile ground and an essential prerequisite for selectively targeting metal-binding proteins with small-molecule inhibitors aiming to (i) abrogate deregulated metal-dependent (mis)functions or (ii) leverage metal-dyshomeostasis to selectively trigger harmful cells death.
Collapse
Affiliation(s)
- Angelo Spinello
- National Research Council of Italy (CNR)-, Institute of Materials (IOM) c/o International School for Advanced Studies (SISSA), via Bonomea 265, 34136, Trieste, Italy
| | - Jure Borišek
- National Institute of Chemistry Institution Hajdrihova ulica 19, 1000, Ljubljana, Slovenia
| | - Matic Pavlin
- Laboratory of Microsensor Structures and Electronics Faculty of Electrical Engineering, University of Ljubljana Tržaška cesta 25, 1000, Ljubljana, Slovenia
| | - Pavel Janoš
- National Research Council of Italy (CNR)-, Institute of Materials (IOM) c/o International School for Advanced Studies (SISSA), via Bonomea 265, 34136, Trieste, Italy
| | - Alessandra Magistrato
- National Research Council of Italy (CNR)-, Institute of Materials (IOM) c/o International School for Advanced Studies (SISSA), via Bonomea 265, 34136, Trieste, Italy
| |
Collapse
|
18
|
Fischer A, Smieško M, Sellner M, Lill MA. Decision Making in Structure-Based Drug Discovery: Visual Inspection of Docking Results. J Med Chem 2021; 64:2489-2500. [PMID: 33617246 DOI: 10.1021/acs.jmedchem.0c02227] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Molecular docking is a computational method widely used in drug discovery. Due to the inherent inaccuracies of molecular docking, visual inspection of binding modes is a crucial routine in the decision making process of computational medicinal chemists. Despite its apparent importance for medicinal chemistry projects, guidelines for the visual docking pose assessment have been hardly discussed in the literature. Here, we review the medicinal chemistry literature with the aim of identifying consistent principles for visual inspection, highlighting cases of its successful application, and discussing its limitations. In this context, we conducted a survey reaching experts in both academia and the pharmaceutical industry, which also included a challenge to distinguish native from incorrect poses. We were able to collect 93 expert opinions that offer valuable insights into visually supported decision-making processes. This perspective shall motivate discussions among experienced computational medicinal chemists and guide young scientists new to the field to stratify their compounds.
Collapse
Affiliation(s)
- André Fischer
- Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland
| | - Martin Smieško
- Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland
| | - Manuel Sellner
- Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland
| | - Markus A Lill
- Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland
| |
Collapse
|
19
|
Microwave-assisted synthesis, biological assessment, and molecular modeling of aza-heterocycles: Potential inhibitory capacity of cholinergic enzymes to Alzheimer's disease. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Ammazzalorso A, Gallorini M, Fantacuzzi M, Gambacorta N, De Filippis B, Giampietro L, Maccallini C, Nicolotti O, Cataldi A, Amoroso R. Design, synthesis and biological evaluation of imidazole and triazole-based carbamates as novel aromatase inhibitors. Eur J Med Chem 2020; 211:113115. [PMID: 33360796 DOI: 10.1016/j.ejmech.2020.113115] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/07/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Abstract
In the search for novel aromatase inhibitors, a series of triazole and imidazole-based carbamate derivatives were designed and synthesized. Final compounds were thus evaluated against human aromatase by in vitro kinetic experiments in a fluorimetric assay in comparison with letrozole. The effect of most active derivatives 13a and 15c was then evaluated in vitro on the human breast cancer cell line MCF7 by MTT assay, cytotoxicity assay (LDH release) and cell cycle analysis, revealing a dose-dependent inhibition profile of cell viability and low micromolar IC50 values. In addition, docking simulations were also carried out to elucidate at a molecular level of detail the binding modes adopted to target human aromatase.
Collapse
Affiliation(s)
- Alessandra Ammazzalorso
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy.
| | - Marialucia Gallorini
- Unit of Anatomy, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Marialuigia Fantacuzzi
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Nicola Gambacorta
- Unit of Medicinal Chemistry, Department of Farmacia-Scienze Del Farmaco, "A. Moro" University, Bari, Italy
| | - Barbara De Filippis
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Letizia Giampietro
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Cristina Maccallini
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Orazio Nicolotti
- Unit of Medicinal Chemistry, Department of Farmacia-Scienze Del Farmaco, "A. Moro" University, Bari, Italy
| | - Amelia Cataldi
- Unit of Anatomy, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Rosa Amoroso
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| |
Collapse
|
21
|
Caciolla J, Spinello A, Martini S, Bisi A, Zaffaroni N, Gobbi S, Magistrato A. Targeting Orthosteric and Allosteric Pockets of Aromatase via Dual-Mode Novel Azole Inhibitors. ACS Med Chem Lett 2020; 11:732-739. [PMID: 32435378 DOI: 10.1021/acsmedchemlett.9b00591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Breast cancer (BC) is the most diffused cancer type in women and the second leading cause of death among the female population. Effective strategies to fight estrogen responsive (ER+) BC, which represents 70% of all BC cases, rely on estrogen deprivation, via the inhibition of the aromatase enzyme, or the modulation of its cognate estrogen receptor. Current clinical therapies significantly increased patient survival time. Nevertheless, the onset of resistance in metastatic BC patients undergoing prolonged treatments is becoming a current clinical challenge, urgently demanding to devise innovative strategies. In this context, here we designed, synthesized, and performed in vitro inhibitory tests on the aromatase enzyme and distinct ER+/ER- BC cell line types of novel azole bridged xanthones. These compounds are active in the low μM range and behave as dual-mode inhibitors, targeting both the orthosteric and the allosteric sites of the enzyme placed along one access channel. Classical and quantum-classical molecular dynamics simulations of the new compounds, as compared with selected steroidal and nonsteroidal inhibitors, provide a rationale to the observed inhibitory potency and supply the guidelines to boost the activity of inhibitors able to exploit coordination to iron and occupation of the access channel to modulate estrogen production.
Collapse
Affiliation(s)
- Jessica Caciolla
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Angelo Spinello
- CNR-IOM Democritos c/o International School for Advanced Studies (SISSA), Via Bonomea 265, 34136 Trieste, Italy
| | - Silvia Martini
- Fondazione IRCSS Istituto Nazionale dei Tumori, via Amadeo 42, 20113 Milano, Italy
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Nadia Zaffaroni
- Fondazione IRCSS Istituto Nazionale dei Tumori, via Amadeo 42, 20113 Milano, Italy
| | - Silvia Gobbi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Alessandra Magistrato
- CNR-IOM Democritos c/o International School for Advanced Studies (SISSA), Via Bonomea 265, 34136 Trieste, Italy
| |
Collapse
|
22
|
Fantacuzzi M, De Filippis B, Gallorini M, Ammazzalorso A, Giampietro L, Maccallini C, Aturki Z, Donati E, Ibrahim RS, Shawky E, Cataldi A, Amoroso R. Synthesis, biological evaluation, and docking study of indole aryl sulfonamides as aromatase inhibitors. Eur J Med Chem 2020; 185:111815. [DOI: 10.1016/j.ejmech.2019.111815] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/30/2022]
|
23
|
Novel triazole-tetrahydroisoquinoline hybrids as human aromatase inhibitors. Bioorg Chem 2019; 93:103327. [DOI: 10.1016/j.bioorg.2019.103327] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/14/2019] [Accepted: 09/27/2019] [Indexed: 01/06/2023]
|
24
|
Adenosine Derivates as Antioxidant Agents: Synthesis, Characterization, in Vitro Activity, and Theoretical Insights. Antioxidants (Basel) 2019; 8:antiox8100468. [PMID: 31600955 PMCID: PMC6826950 DOI: 10.3390/antiox8100468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/25/2019] [Accepted: 09/29/2019] [Indexed: 11/17/2022] Open
Abstract
In this work, we present results about the synthesis and the antioxidant properties of seven adenosine derivatives. Four of these compounds were synthesized by substituting the N6-position of adenosine with aliphatic amines, and three were obtained by modification of the ribose ring. All compounds were obtained in pure form using column chromatography, and their structures were elucidated by infrared spectroscopy (IR) and Nuclear Magnetic Resonance (NMR). All adenosine derivatives were further evaluated in vitro as free radical scavengers. Our results show that compounds 1c, 3, and 5 display a potent antioxidant effect compared with the reference compound ascorbic acid. In addition, the absorption, distribution, metabolism and excretion (ADME) calculations show favorable pharmacokinetic parameters for the set of compounds analyzed, which guarantees their suitability as potential antioxidant drugs. Furthermore, theoretical analyses using Molecular Quantum Similarity and reactivity indices were performed in order to discriminate the different reactive sites involved in oxidative processes.
Collapse
|
25
|
Preto J, Gentile F. Assessing and improving the performance of consensus docking strategies using the DockBox package. J Comput Aided Mol Des 2019; 33:817-829. [DOI: 10.1007/s10822-019-00227-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
|
26
|
Leechaisit R, Pingaew R, Prachayasittikul V, Worachartcheewan A, Prachayasittikul S, Ruchirawat S, Prachayasittikul V. Synthesis, molecular docking, and QSAR study of bis-sulfonamide derivatives as potential aromatase inhibitors. Bioorg Med Chem 2019; 27:115040. [DOI: 10.1016/j.bmc.2019.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 01/03/2023]
|
27
|
Discovery of Novel TASK-3 Channel Blockers Using a Pharmacophore-Based Virtual Screening. Int J Mol Sci 2019; 20:ijms20164014. [PMID: 31426491 PMCID: PMC6720600 DOI: 10.3390/ijms20164014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/06/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023] Open
Abstract
TASK-3 is a two-pore domain potassium (K2P) channel highly expressed in the hippocampus, cerebellum, and cortex. TASK-3 has been identified as an oncogenic potassium channel and it is overexpressed in different cancer types. For this reason, the development of new TASK-3 blockers could influence the pharmacological treatment of cancer and several neurological conditions. In the present work, we searched for novel TASK-3 blockers by using a virtual screening protocol that includes pharmacophore modeling, molecular docking, and free energy calculations. With this protocol, 19 potential TASK-3 blockers were identified. These molecules were tested in TASK-3 using patch clamp, and one blocker (DR16) was identified with an IC50 = 56.8 ± 3.9 μM. Using DR16 as a scaffold, we designed DR16.1, a novel TASK-3 inhibitor, with an IC50 = 14.2 ± 3.4 μM. Our finding takes on greater relevance considering that not many inhibitory TASK-3 modulators have been reported in the scientific literature until today. These two novel TASK-3 channel inhibitors (DR16 and DR16.1) are the first compounds found using a pharmacophore-based virtual screening and rational drug design protocol.
Collapse
|
28
|
Rodriguez Núñez YA, Norambuena M, Romero Bohorquez AR, Morales-Bayuelo A, Gutíerrez M. Efficient synthesis and antioxidant activity of novel N-propargyl tetrahydroquinoline derivatives through the cationic Povarov reaction. Heliyon 2019; 5:e02174. [PMID: 31417970 PMCID: PMC6690562 DOI: 10.1016/j.heliyon.2019.e02174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/17/2019] [Accepted: 07/25/2019] [Indexed: 12/31/2022] Open
Abstract
New N-propargyl tetrahydroquinolines 6a-g have been synthesized efficiently through the cationic Povarov reaction (a domino Mannich/Friedel-Crafts reaction), catalyzed by Indium (III) chloride (InCl3), from the corresponding N-propargylanilines preformed, formaldehyde and N-vinylformamide, with good to moderate yields. All tetrahydroquinoline derivatives obtained were evaluated in vitro as free radical scavengers. Results showed that compound 6c presents a potent antioxidant effect compared with ascorbic acid, used as a reference compound. ADME predictions also revealed favorable pharmacokinetic parameters for the synthesized compounds, which warrant their suitability as potentials antioxidant. Additionally, a theoretical study using Molecular Quantum Similarity and reactivity indices were developed to discriminate different reactive sites in the new molecules in which the oxidative process occurs.
Collapse
Affiliation(s)
- Yeray A. Rodriguez Núñez
- Laboratorio Síntesis Orgánica, Instituto de Química de Recursos Naturales, Universidad de Talca, Casilla 747, Talca, 3460000, Chile
| | - Maximiliano Norambuena
- Laboratorio Síntesis Orgánica, Instituto de Química de Recursos Naturales, Universidad de Talca, Casilla 747, Talca, 3460000, Chile
| | - Arnold R. Romero Bohorquez
- Grupo de Investigación de Compuestos Orgánicos de Interés Medicinal (CODEIM), Parque Tecnológico Guatiguará, Universidad Industrial de Santander, A.A. 678, Piedecuesta, Colombia
| | - Alejandro Morales-Bayuelo
- Centro de Investigación de Procesos del Tecnologico Comfenalco (CIPTEC), programa de Ingeniería Industrial, Fundacion Universitaria Tecnologico Comfenalco – Cartagena, Cr 44 D N 30A, 91, Cartagena-Bolívar, Colombia
| | - Margarita Gutíerrez
- Laboratorio Síntesis Orgánica, Instituto de Química de Recursos Naturales, Universidad de Talca, Casilla 747, Talca, 3460000, Chile
| |
Collapse
|
29
|
Spinello A, Ritacco I, Magistrato A. Recent advances in computational design of potent aromatase inhibitors: open-eye on endocrine-resistant breast cancers. Expert Opin Drug Discov 2019; 14:1065-1076. [DOI: 10.1080/17460441.2019.1646245] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Angelo Spinello
- National Research Council - Istituto Officina dei Materiali c/o International School for Advanced Studies (SISSA), Trieste, Italy
| | - Ida Ritacco
- National Research Council - Istituto Officina dei Materiali c/o International School for Advanced Studies (SISSA), Trieste, Italy
| | - Alessandra Magistrato
- National Research Council - Istituto Officina dei Materiali c/o International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
30
|
Liu XG, Lv MC, Huang MY, Sun YQ, Gao PY, Li DQ. A network pharmacology study on the triterpene saponins from Medicago sativa L. for the treatment of Neurodegenerative diseases. J Food Biochem 2019; 43:e12955. [PMID: 31368545 DOI: 10.1111/jfbc.12955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 12/26/2022]
Abstract
Neurodegenerative diseases (NDDs) are characterized by progressive and irreversible, is a kind of complex illnesses, and the long-term therapy which is frequently associated with adverse side effects. Medicago sativa L., widely consumed as a vegetable, has the effects of improving memory and relieving central nervous system diseases. However, there are less studies on its specific mechanism for NDDs. In this investigation, we applied a method of network pharmacology, which combined molecular docking and network analysis to decipher the mechanisms of M. sativa in NDDs. The pharmacological system generated 55 triterpene saponins from M. sativa, and predicted 27 potential targets with 100 pathways in the treatment of NDDs. As a result, 13 compounds, 10 target proteins, and 6 signaling pathways were found to play important roles in the treatment of NDDs. In addition, in vitro experiments of isolates confirmed activities for NDDs, which were consistent with the results of network pharmacology prediction. PRACTICAL APPLICATIONS: Medicago sativa L. has been widely consumed as a vegetable, which possesses many nutritional components. As a functional food stuff, M. sativa can improve human health, such as memory improving activities, relieving central nervous system diseases, immunomodulatory, antioxidant, anticancer, and anti-inflammatory. In this article, the mechanism of triterpene saponins from M. sativa against NDDs was successfully predicted by network pharmacology method. The results will serve as a reference of M. sativa against NDDs.
Collapse
Affiliation(s)
- Xue-Gui Liu
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, P.R. China.,Institute of Functional Molecules, Shenyang University of Chemical Technology, Shenyang, P.R. China
| | - Meng-Chao Lv
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, P.R. China
| | - Ming-Yuan Huang
- Shenyang Institute of Science and Technology, Shenyang, P.R. China
| | - Yu-Qiu Sun
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, P.R. China
| | - Pin-Yi Gao
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, P.R. China.,Institute of Functional Molecules, Shenyang University of Chemical Technology, Shenyang, P.R. China
| | - Dan-Qi Li
- Institute of Functional Molecules, Shenyang University of Chemical Technology, Shenyang, P.R. China
| |
Collapse
|
31
|
Rendic SP, Peter Guengerich F. Human cytochrome P450 enzymes 5-51 as targets of drugs and natural and environmental compounds: mechanisms, induction, and inhibition - toxic effects and benefits. Drug Metab Rev 2019; 50:256-342. [PMID: 30717606 DOI: 10.1080/03602532.2018.1483401] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 (P450, CYP) enzymes have long been of interest due to their roles in the metabolism of drugs, pesticides, pro-carcinogens, and other xenobiotic chemicals. They have also been of interest due to their very critical roles in the biosynthesis and metabolism of steroids, vitamins, and certain eicosanoids. This review covers the 22 (of the total of 57) human P450s in Families 5-51 and their substrate selectivity. Furthermore, included is information and references regarding inducibility, inhibition, and (in some cases) stimulation by chemicals. We update and discuss important aspects of each of these 22 P450s and questions that remain open.
Collapse
Affiliation(s)
| | - F Peter Guengerich
- b Department of Biochemistry , Vanderbilt University School of Medicine , Nashville , TN , USA
| |
Collapse
|
32
|
Can multiscale simulations unravel the function of metallo-enzymes to improve knowledge-based drug discovery? Future Med Chem 2019; 11:771-791. [DOI: 10.4155/fmc-2018-0495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Metallo-enzymes are a large class of biomolecules promoting specialized chemical reactions. Quantum-classical quantum mechanics/molecular mechanics molecular dynamics, describing the metal site at quantum mechanics level, while accounting for the rest of system at molecular mechanics level, has an accessible time-scale limited by its computational cost. Hence, it must be integrated with classical molecular dynamics and enhanced sampling simulations to disentangle the functions of metallo-enzymes. In this review, we provide an overview of these computational methods and their capabilities. In particular, we will focus on some systems such as CYP19A1 a Fe-dependent enzyme involved in estrogen biosynthesis, and on Mg2+-dependent DNA/RNA processing enzymes/ribozymes and the spliceosome, a protein-directed ribozyme. This information may guide the discovery of drug-like molecules and genetic manipulation tools.
Collapse
|
33
|
Spinello A, Martini S, Berti F, Pennati M, Pavlin M, Sgrignani J, Grazioso G, Colombo G, Zaffaroni N, Magistrato A. Rational design of allosteric modulators of the aromatase enzyme: An unprecedented therapeutic strategy to fight breast cancer. Eur J Med Chem 2019; 168:253-262. [DOI: 10.1016/j.ejmech.2019.02.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 12/29/2022]
|
34
|
Roleira FMF, Varela C, Amaral C, Costa SC, Correia-da-Silva G, Moraca F, Costa G, Alcaro S, Teixeira NAA, Tavares da Silva EJ. C-6α- vs C-7α-Substituted Steroidal Aromatase Inhibitors: Which Is Better? Synthesis, Biochemical Evaluation, Docking Studies, and Structure–Activity Relationships. J Med Chem 2019; 62:3636-3657. [DOI: 10.1021/acs.jmedchem.9b00157] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Fernanda M. F. Roleira
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- CIEPQPF Centre for Chemical Processes Engineering and Forest Products, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Carla Varela
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- CIEPQPF Centre for Chemical Processes Engineering and Forest Products, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Cristina Amaral
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Saul C. Costa
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Federica Moraca
- Laboratorio di Chimica Farmaceutica, Dipartimento di Scienze della Salute, Università Magna Græcia di Catanzaro, 88100 Catanzaro, Italy
- Department of Pharmacy, University of Naples “Federico II”, via D. Montesano 49, 80131, Naples, Italy
- Net4Science Academic Spin-Off, “Magna Græcia” University of Catanzaro, “S. Venuta”, Catanzaro, Italy
| | - Giosuè Costa
- Laboratorio di Chimica Farmaceutica, Dipartimento di Scienze della Salute, Università Magna Græcia di Catanzaro, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, “Magna Græcia” University of Catanzaro, “S. Venuta”, Catanzaro, Italy
| | - Stefano Alcaro
- Laboratorio di Chimica Farmaceutica, Dipartimento di Scienze della Salute, Università Magna Græcia di Catanzaro, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, “Magna Græcia” University of Catanzaro, “S. Venuta”, Catanzaro, Italy
| | - Natércia A. A. Teixeira
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Elisiário J. Tavares da Silva
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- CIEPQPF Centre for Chemical Processes Engineering and Forest Products, University of Coimbra, 3030-790 Coimbra, Portugal
| |
Collapse
|
35
|
The Catalytic Mechanism of Steroidogenic Cytochromes P450 from All-Atom Simulations: Entwinement with Membrane Environment, Redox Partners, and Post-Transcriptional Regulation. Catalysts 2019. [DOI: 10.3390/catal9010081] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cytochromes P450 (CYP450s) promote the biosynthesis of steroid hormones with major impact on the onset of diseases such as breast and prostate cancers. By merging distinct functions into the same catalytic scaffold, steroidogenic CYP450s enhance complex chemical transformations with extreme efficiency and selectivity. Mammalian CYP450s and their redox partners are membrane-anchored proteins, dynamically associating to form functional machineries. Mounting evidence signifies that environmental factors are strictly intertwined with CYP450s catalysis. Atomic-level simulations have the potential to provide insights into the catalytic mechanism of steroidogenic CYP450s and on its regulation by environmental factors, furnishing information often inaccessible to experimental means. In this review, after an introduction of computational methods commonly employed to tackle these systems, we report the current knowledge on three steroidogenic CYP450s—CYP11A1, CYP17A1, and CYP19A1—endowed with multiple catalytic functions and critically involved in cancer onset. In particular, besides discussing their catalytic mechanisms, we highlight how the membrane environment contributes to (i) regulate ligand channeling through these enzymes, (ii) modulate their interactions with specific protein partners, (iii) mediate post-transcriptional regulation induced by phosphorylation. The results presented set the basis for developing novel therapeutic strategies aimed at fighting diseases originating from steroid metabolism dysfunction.
Collapse
|
36
|
Forero Doria O, Castro R, Gutierrez M, Gonzalez Valenzuela D, Santos L, Ramirez D, Guzman L. Novel Alkylimidazolium Ionic Liquids as an Antibacterial Alternative to Pathogens of the Skin and Soft Tissue Infections. Molecules 2018; 23:molecules23092354. [PMID: 30223457 PMCID: PMC6225289 DOI: 10.3390/molecules23092354] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/03/2018] [Accepted: 09/10/2018] [Indexed: 01/05/2023] Open
Abstract
Keeping in mind the concept of green chemistry, this research aims to synthesize and characterize new ionic liquids (ILs) derived from N-cinnamyl imidazole with different sizes of alkyl chains (1, 6, 8, and 10 carbon atoms), and evaluate their antibacterial activity against Skin and soft tissue infections (SSTIs) causative bacteria. The antibacterial screening was carried out by agar well diffusion and the Minimum Inhibitory Concentration (MIC) and Half Maximum Inhibitory Concentration (IC50) of the different ILs were determined by microdilution in broth, also Molecular dynamics simulations were performed to study the interaction mechanism between ILs and membranes. The MIC value in Gram-positive bacteria showed that as the hydrocarbon chain increases, the MIC value decreases with a dose-dependent effect. Furthermore, Gram-negative bacteria showed high MIC values, which were also evidenced in the antibacterial screening. The molecular dynamics showed an incorporation of the ILs with the longer chain (10 C), corresponding to a passive diffusion towards the membrane surface, for its part, the ILs with the shorter chain due to its lack of hydrophobicity was not incorporated into the bilayer. Finally, the new ILs synthesized could be an alternative for the treatment of Gram-positive bacteria causative of SSTIs.
Collapse
Affiliation(s)
- Oscar Forero Doria
- Instituto de Química de Recursos Naturales, Universidad de Talca, P.O. Box 747, Talca 3460000, Chile.
| | - Ricardo Castro
- Multidisciplinary Agroindustry Research Laboratory, Universidad Autónoma de Chile, Talca 3460000, Chile.
- Carrera de Ingeniería en Construcción e Instituto de Ciencias Químicas Aplicadas, Universidad Autónoma de Chile, Talca 3460000, Chile.
| | - Margarita Gutierrez
- Instituto de Química de Recursos Naturales, Universidad de Talca, P.O. Box 747, Talca 3460000, Chile.
| | | | - Leonardo Santos
- Instituto de Química de Recursos Naturales, Universidad de Talca, P.O. Box 747, Talca 3460000, Chile.
| | - David Ramirez
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca 3460000, Chile.
| | - Luis Guzman
- Departamento de Bioquímica Clínica e Inmunohematología, Facultad de Ciencias de la Salud, Universidad de Talca, P.O. Box 747, Talca 3460000, Chile.
| |
Collapse
|
37
|
|
38
|
Sahin Z, Ertas M, Berk B, Biltekin SN, Yurttas L, Demirayak S. Studies on non-steroidal inhibitors of aromatase enzyme; 4-(aryl/heteroaryl)-2-(pyrimidin-2-yl)thiazole derivatives. Bioorg Med Chem 2018. [DOI: 10.1016/j.bmc.2018.02.048] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
39
|
Ramírez D, Caballero J. Is It Reliable to Take the Molecular Docking Top Scoring Position as the Best Solution without Considering Available Structural Data? Molecules 2018; 23:molecules23051038. [PMID: 29710787 PMCID: PMC6102569 DOI: 10.3390/molecules23051038] [Citation(s) in RCA: 262] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/21/2018] [Accepted: 04/26/2018] [Indexed: 12/13/2022] Open
Abstract
Molecular docking is the most frequently used computational method for studying the interactions between organic molecules and biological macromolecules. In this context, docking allows predicting the preferred pose of a ligand inside a receptor binding site. However, the selection of the “best” solution is not a trivial task, despite the widely accepted selection criterion that the best pose corresponds to the best energy score. Here, several rigid-target docking methods were evaluated on the same dataset with respect to their ability to reproduce crystallographic binding orientations, to test if the best energy score is a reliable criterion for selecting the best solution. For this, two experiments were performed: (A) to reconstruct the ligand-receptor complex by performing docking of the ligand in its own crystal structure receptor (defined as self-docking), and (B) to reconstruct the ligand-receptor complex by performing docking of the ligand in a crystal structure receptor that contains other ligand (defined as cross-docking). Root-mean square deviation (RMSD) was used to evaluate how different the obtained docking orientation is from the corresponding co-crystallized pose of the same ligand molecule. We found that docking score function is capable of predicting crystallographic binding orientations, but the best ranked solution according to the docking energy is not always the pose that reproduces the experimental binding orientation. This happened when self-docking was achieved, but it was critical in cross-docking. Taking into account that docking is typically used with predictive purposes, during cross-docking experiments, our results indicate that the best energy score is not a reliable criterion to select the best solution in common docking applications. It is strongly recommended to choose the best docking solution according to the scoring function along with additional structural criteria described for analogue ligands to assure the selection of a correct docking solution.
Collapse
Affiliation(s)
- David Ramírez
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, 5 Poniente No. 1670, 3460000 Talca, Chile.
| | - Julio Caballero
- Centro de Bioinformática y Simulación Molecular (CBSM), Universidad de Talca. 1 Poniente No. 1141, 3460000 Talca, Chile.
| |
Collapse
|
40
|
Ertas M, Sahin Z, Berk B, Yurttas L, Biltekin SN, Demirayak S. Pyridine-substituted thiazolylphenol derivatives: Synthesis, modeling studies, aromatase inhibition, and antiproliferative activity evaluation. Arch Pharm (Weinheim) 2018. [DOI: 10.1002/ardp.201700272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Merve Ertas
- Department of Pharmaceutical Chemistry, School of Pharmacy; İstanbul Medipol University; İstanbul Turkey
| | - Zafer Sahin
- Department of Pharmaceutical Chemistry, School of Pharmacy; İstanbul Medipol University; İstanbul Turkey
| | - Barkin Berk
- Department of Pharmaceutical Chemistry, School of Pharmacy; İstanbul Medipol University; İstanbul Turkey
| | - Leyla Yurttas
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry; Anadolu University; Eskisehir Turkey
| | - Sevde N. Biltekin
- Department of Pharmaceutical Microbiology, School of Pharmacy; İstanbul Medipol University; İstanbul Turkey
| | - Seref Demirayak
- Department of Pharmaceutical Chemistry, School of Pharmacy; İstanbul Medipol University; İstanbul Turkey
| |
Collapse
|
41
|
Karimian Amroabadi M, Taheri-Kafrani A, Heidarpoor Saremi L, Rastegari AA. Spectroscopic studies of the interaction between alprazolam and apo-human serum transferrin as a drug carrier protein. Int J Biol Macromol 2018; 108:263-271. [DOI: 10.1016/j.ijbiomac.2017.11.179] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 01/20/2023]
|
42
|
Prior AM, Yu X, Park EJ, Kondratyuk TP, Lin Y, Pezzuto JM, Sun D. Structure-activity relationships and docking studies of synthetic 2-arylindole derivatives determined with aromatase and quinone reductase 1. Bioorg Med Chem Lett 2017; 27:5393-5399. [PMID: 29153737 PMCID: PMC5705205 DOI: 10.1016/j.bmcl.2017.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 01/02/2023]
Abstract
In our ongoing effort of discovering anticancer and chemopreventive agents, a series of 2-arylindole derivatives were synthesized and evaluated toward aromatase and quinone reductase 1 (QR1). Biological evaluation revealed that several compounds (e.g., 2d, IC50 = 1.61 μM; 21, IC50 = 3.05 μM; and 27, IC50 = 3.34 μM) showed aromatase inhibitory activity with half maximal inhibitory concentration (IC50) values in the low micromolar concentrations. With regard to the QR1 induction activity, 11 exhibited the highest QR1 induction ratio (IR) with a low concentration to double activity (CD) value (IR = 8.34, CD = 2.75 μM), while 7 showed the most potent CD value of 1.12 μM. A dual acting compound 24 showed aromatase inhibition (IC50 = 9.00 μM) as well as QR1 induction (CD = 5.76 μM) activities. Computational docking studies using CDOCKER (Discovery Studio 3.5) provided insight in regard to the potential binding modes of 2-arylindoles within the aromatase active site. Predominantly, the 2-arylindoles preferred binding with the 2-aryl group toward a small hydrophobic pocket within the active site. The C-5 electron withdrawing group on indole was predicted to have an important role and formed a hydrogen bond with Ser478 (OH). Alternatively, meta-pyridyl analogs may orient with the pyridyl 3'-nitrogen coordinating with the heme group.
Collapse
Affiliation(s)
- Allan M Prior
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, 34 Rainbow Drive, Hilo, HI 96720, USA
| | - Xufen Yu
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, 34 Rainbow Drive, Hilo, HI 96720, USA
| | - Eun-Jung Park
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, 34 Rainbow Drive, Hilo, HI 96720, USA; Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA
| | - Tamara P Kondratyuk
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, 34 Rainbow Drive, Hilo, HI 96720, USA
| | - Yan Lin
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, 34 Rainbow Drive, Hilo, HI 96720, USA
| | - John M Pezzuto
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, 34 Rainbow Drive, Hilo, HI 96720, USA; Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA
| | - Dianqing Sun
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, 34 Rainbow Drive, Hilo, HI 96720, USA.
| |
Collapse
|
43
|
Adhikari N, Amin SA, Saha A, Jha T. Combating breast cancer with non-steroidal aromatase inhibitors (NSAIs): Understanding the chemico-biological interactions through comparative SAR/QSAR study. Eur J Med Chem 2017. [DOI: 10.1016/j.ejmech.2017.05.041] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
44
|
Du H, Cai Y, Yang H, Zhang H, Xue Y, Liu G, Tang Y, Li W. In Silico Prediction of Chemicals Binding to Aromatase with Machine Learning Methods. Chem Res Toxicol 2017; 30:1209-1218. [PMID: 28414904 DOI: 10.1021/acs.chemrestox.7b00037] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Environmental chemicals may affect endocrine systems through multiple mechanisms, one of which is via effects on aromatase (also known as CYP19A1), an enzyme critical for maintaining the normal balance of estrogens and androgens in the body. Therefore, rapid and efficient identification of aromatase-related endocrine disrupting chemicals (EDCs) is important for toxicology and environment risk assessment. In this study, on the basis of the Tox21 10K compound library, in silico classification models for predicting aromatase binders/nonbinders were constructed by machine learning methods. To improve the prediction ability of the models, a combined classifier (CC) strategy that combines different independent machine learning methods was adopted. Performances of the models were measured by test and external validation sets containing 1336 and 216 chemicals, respectively. The best model was obtained with the MACCS (Molecular Access System) fingerprint and CC method, which exhibited an accuracy of 0.84 for the test set and 0.91 for the external validation set. Additionally, several representative substructures for characterizing aromatase binders, such as ketone, lactone, and nitrogen-containing derivatives, were identified using information gain and substructure frequency analysis. Our study provided a systematic assessment of chemicals binding to aromatase. The built models can be helpful to rapidly identify potential EDCs targeting aromatase.
Collapse
Affiliation(s)
- Hanwen Du
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Yingchun Cai
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Hongbin Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Hongxiao Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Yuhan Xue
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| |
Collapse
|
45
|
Uehara S, Tanaka S. Cosolvent-Based Molecular Dynamics for Ensemble Docking: Practical Method for Generating Druggable Protein Conformations. J Chem Inf Model 2017; 57:742-756. [PMID: 28388074 DOI: 10.1021/acs.jcim.6b00791] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Protein flexibility is a major hurdle in current structure-based virtual screening (VS). In spite of the recent advances in high-performance computing, protein-ligand docking methods still demand tremendous computational cost to take into account the full degree of protein flexibility. In this context, ensemble docking has proven its utility and efficiency for VS studies, but it still needs a rational and efficient method to select and/or generate multiple protein conformations. Molecular dynamics (MD) simulations are useful to produce distinct protein conformations without abundant experimental structures. In this study, we present a novel strategy that makes use of cosolvent-based molecular dynamics (CMD) simulations for ensemble docking. By mixing small organic molecules into a solvent, CMD can stimulate dynamic protein motions and induce partial conformational changes of binding pocket residues appropriate for the binding of diverse ligands. The present method has been applied to six diverse target proteins and assessed by VS experiments using many actives and decoys of DEKOIS 2.0. The simulation results have revealed that the CMD is beneficial for ensemble docking. Utilizing cosolvent simulation allows the generation of druggable protein conformations, improving the VS performance compared with the use of a single experimental structure or ensemble docking by standard MD with pure water as the solvent.
Collapse
Affiliation(s)
- Shota Uehara
- Department of Computational Science, Graduate School of System Informatics, Kobe University , 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Shigenori Tanaka
- Department of Computational Science, Graduate School of System Informatics, Kobe University , 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| |
Collapse
|
46
|
Mojaddami A, Sakhteman A, Fereidoonnezhad M, Faghih Z, Najdian A, Khabnadideh S, Sadeghpour H, Rezaei Z. Binding mode of triazole derivatives as aromatase inhibitors based on docking, protein ligand interaction fingerprinting, and molecular dynamics simulation studies. Res Pharm Sci 2017; 12:21-30. [PMID: 28255310 PMCID: PMC5333476 DOI: 10.4103/1735-5362.199043] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Aromatase inhibitors (AIs) as effective candidates have been used in the treatment of hormone-dependent breast cancer. In this study, we have proposed 300 structures as potential AIs and filtered them by Lipinski's rule of five using DrugLito software. Subsequently, they were subjected to docking simulation studies to select the top 20 compounds based on their Gibbs free energy changes and also to perform more studies on the protein-ligand interaction fingerprint by AuposSOM software. In this stage, anastrozole and letrozole were used as positive control to compare their interaction fingerprint patterns with our proposed structures. Finally, based on the binding energy values, one active structure (ligand 15) was selected for molecular dynamic simulation in order to get information for the binding mode of these ligands within the enzyme cavity. The triazole of ligand 15 pointed to HEM group in aromatase active site and coordinated to Fe of HEM through its N4 atom. In addition, two π-cation interactions was also observed, one interaction between triazole and porphyrin of HEM group, and the other was 4-chloro phenyl moiety of this ligand with Arg115 residue.
Collapse
Affiliation(s)
- Ayyub Mojaddami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Centre, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, I.R. Iran
| | - Amirhossein Sakhteman
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Centre, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, I.R. Iran
| | - Masood Fereidoonnezhad
- Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, I.R. Iran
| | - Zeinab Faghih
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Centre, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, I.R. Iran
| | - Atena Najdian
- Department of Nuclear Pharmacy, School of Pharmacy, Tehran University of Medical Sciences, Tehran, I.R. Iran
| | - Soghra Khabnadideh
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Centre, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, I.R. Iran
| | - Hossein Sadeghpour
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Centre, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, I.R. Iran
| | - Zahra Rezaei
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Centre, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, I.R. Iran
| |
Collapse
|
47
|
Sobral AF, Amaral C, Correia-da-Silva G, Teixeira N. Unravelling exemestane: From biology to clinical prospects. J Steroid Biochem Mol Biol 2016; 163:1-11. [PMID: 26992705 DOI: 10.1016/j.jsbmb.2016.03.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/29/2016] [Accepted: 03/13/2016] [Indexed: 11/28/2022]
Abstract
Aromatase inhibitors (AIs) are anti-tumor agents used in clinic to treat hormone-dependent breast cancer. AIs block estrogens biosynthesis by inhibiting the enzyme aromatase, preventing tumor progression. Exemestane, a third-generation steroidal AI, belongs to this class of drugs and is currently used in clinic to treat postmenopausal women, due to its high efficacy and good tolerability. Here, its pharmacological and biological aspects as well as its clinical applications and comparison to other endocrine therapeutic agents, are reviewed. It is also focused the benefits and risks of exemestane, drawbacks to be overcome and aspects to be explored.
Collapse
Affiliation(s)
- Ana Filipa Sobral
- Faculty of Science and Technology, University of Coimbra, Calçada Martim de Freitas 3000-456 Coimbra, Portugal; UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal
| | - Cristina Amaral
- UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal.
| | - Georgina Correia-da-Silva
- UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal
| | - Natércia Teixeira
- UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal.
| |
Collapse
|
48
|
Navarro-Retamal C, Caballero J. Flavonoids as CDK1 Inhibitors: Insights in Their Binding Orientations and Structure-Activity Relationship. PLoS One 2016; 11:e0161111. [PMID: 27517610 PMCID: PMC4982677 DOI: 10.1371/journal.pone.0161111] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/29/2016] [Indexed: 01/20/2023] Open
Abstract
In the last years, the interactions of flavonoids with protein kinases (PKs) have been described by using crystallographic experiments. Interestingly, different orientations have been found for one flavonoid inside different PKs and different chemical substitutions lead to different orientations of the flavonoid scaffold inside one PK. Accordingly, orientation predictions of novel analogues could help to the design of flavonoids with high PK inhibitory activities. With this in mind, we studied the binding modes of 37 flavonoids (flavones and chalcones) inside the cyclin-dependent PK CDK1 using docking experiments. We found that the compounds under study adopted two different orientations into the active site of CDK1 (orientations I and II in the manuscript). In addition, quantitative structure-activity relationship (QSAR) models using CoMFA and CoMSIA methodologies were constructed to explain the trend of the CDK1 inhibitory activities for the studied flavonoids. Template-based and docking-based alignments were used. Models developed starting from docking-based alignment were applied for describing the whole dataset and compounds with orientation I. Adequate R2 and Q2 values were obtained by each method; interestingly, only hydrophobic and hydrogen bond donor fields describe the differential potency of the flavonoids as CDK1 inhibitors for both defined alignments and subsets. Our current application of docking and QSAR together reveals important elements to be drawn for the design of novel flavonoids with increased PK inhibitory activities.
Collapse
Affiliation(s)
- Carlos Navarro-Retamal
- Centro de Bioinformática y Simulación Molecular, Facultad de Ingeniería, Universidad de Talca, 2 Norte 685, Casilla 721, Talca, Chile
| | - Julio Caballero
- Centro de Bioinformática y Simulación Molecular, Facultad de Ingeniería, Universidad de Talca, 2 Norte 685, Casilla 721, Talca, Chile
- * E-mail:
| |
Collapse
|
49
|
Bonomo S, Hansen CH, Petrunak EM, Scott EE, Styrishave B, Jørgensen FS, Olsen L. Promising Tools in Prostate Cancer Research: Selective Non-Steroidal Cytochrome P450 17A1 Inhibitors. Sci Rep 2016; 6:29468. [PMID: 27406023 PMCID: PMC4942611 DOI: 10.1038/srep29468] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/20/2016] [Indexed: 01/12/2023] Open
Abstract
Cytochrome P450 17A1 (CYP17A1) is an important target in the treatment of prostate cancer because it produces androgens required for tumour growth. The FDA has approved only one CYP17A1 inhibitor, abiraterone, which contains a steroidal scaffold similar to the endogenous CYP17A1 substrates. Abiraterone is structurally similar to the substrates of other cytochrome P450 enzymes involved in steroidogenesis, and interference can pose a liability in terms of side effects. Using non-steroidal scaffolds is expected to enable the design of compounds that interact more selectively with CYP17A1. Therefore, we combined a structure-based virtual screening approach with density functional theory (DFT) calculations to suggest non-steroidal compounds selective for CYP17A1. In vitro assays demonstrated that two such compounds selectively inhibited CYP17A1 17α-hydroxylase and 17,20-lyase activities with IC50 values in the nanomolar range, without affinity for the major drug-metabolizing CYP2D6 and CYP3A4 enzymes and CYP21A2, with the latter result confirmed in human H295R cells.
Collapse
Affiliation(s)
- Silvia Bonomo
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100, Copenhagen Ø, Denmark
| | - Cecilie H. Hansen
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Elyse M. Petrunak
- Department of Medicinal Chemistry, The University of Kansas, 1251 Wescoe Hall Dr., Lawrence, KS, 66045 USA
| | - Emily E. Scott
- Department of Medicinal Chemistry, The University of Kansas, 1251 Wescoe Hall Dr., Lawrence, KS, 66045 USA
| | - Bjarne Styrishave
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Flemming Steen Jørgensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100, Copenhagen Ø, Denmark
| | - Lars Olsen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100, Copenhagen Ø, Denmark
| |
Collapse
|
50
|
Di Matteo M, Ammazzalorso A, Andreoli F, Caffa I, De Filippis B, Fantacuzzi M, Giampietro L, Maccallini C, Nencioni A, Parenti MD, Soncini D, Del Rio A, Amoroso R. Synthesis and biological characterization of 3-(imidazol-1-ylmethyl)piperidine sulfonamides as aromatase inhibitors. Bioorg Med Chem Lett 2016; 26:3192-3194. [DOI: 10.1016/j.bmcl.2016.04.078] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 10/21/2022]
|